Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 12(1): 3185, 2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-34045461

RESUMEN

Systemic inflammation as manifested in sepsis is an excessive, life-threatening inflammatory response to severe bacterial or viral infection or extensive injury. It is also a thrombo-inflammatory condition associated with vascular leakage/hemorrhage and thrombosis that is not effectively treated by current anti-inflammatory or anti-thrombotic drugs. Here, we show that MB2mP6 peptide nanoparticles, targeting the Gα13-mediated integrin "outside-in" signaling in leukocytes and platelets, inhibited both inflammation and thrombosis without causing hemorrhage/vascular leakage. MB2mP6 improved mouse survival when infused immediately or hours after onset of severe sepsis. Furthermore, platelet Gα13 knockout inhibited septic thrombosis whereas leukocyte Gα13 knockout diminished septic inflammation, each moderately improving survival. Dual platelet/leukocyte Gα13 knockout inhibited septic thrombosis and inflammation, further improving survival similar to MB2mP6. These results demonstrate that inflammation and thrombosis independently contribute to poor outcomes and exacerbate each other in systemic inflammation, and reveal a concept of dual anti-inflammatory/anti-thrombotic therapy without exacerbating vascular leakage.


Asunto(s)
Antígenos CD18/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP G12-G13/antagonistas & inhibidores , Fragmentos de Péptidos/farmacología , Sepsis/tratamiento farmacológico , Trombosis/tratamiento farmacológico , Animales , Antiinflamatorios , Plaquetas/efectos de los fármacos , Plaquetas/inmunología , Plaquetas/metabolismo , Antígenos CD18/metabolismo , Cloruros/administración & dosificación , Cloruros/toxicidad , Modelos Animales de Enfermedad , Compuestos Férricos/administración & dosificación , Compuestos Férricos/toxicidad , Fibrinolíticos , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Humanos , Leucocitos/efectos de los fármacos , Leucocitos/inmunología , Leucocitos/metabolismo , Macrófagos , Ratones , Ratones Noqueados , Nanopartículas/uso terapéutico , Fragmentos de Péptidos/uso terapéutico , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Cultivo Primario de Células , Unión Proteica/efectos de los fármacos , Sepsis/sangre , Sepsis/complicaciones , Sepsis/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Células THP-1 , Trombosis/sangre , Trombosis/inducido químicamente
2.
Circulation ; 138(21): 2395-2412, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-29991487

RESUMEN

BACKGROUND: Although the role of thrombin in atherothrombosis is well studied, its role in the pathogenesis of diet-induced atherosclerosis is not known. METHODS: Using a mouse model of diet-induced atherosclerosis and molecular biological approaches, here we have explored the role of thrombin and its G protein-coupled receptor signaling in diet-induced atherosclerosis. RESULTS: In exploring the role of G protein-coupled receptor signaling in atherogenesis, we found that thrombin triggers foam cell formation via inducing CD36 expression, and these events require Par1-mediated Gα12-Pyk2-Gab1-protein kinase C (PKC)θ-dependent ATF2 activation. Genetic deletion of PKCθ in apolipoprotein E (ApoE)-/- mice reduced Western diet-induced plaque formation. Furthermore, thrombin induced Pyk2, Gab1, PKCθ, and ATF2 phosphorylation, CD36 expression, and foam cell formation in peritoneal macrophages of ApoE-/- mice. In contrast, thrombin only stimulated Pyk2 and Gab1 but not ATF2 phosphorylation or its target gene CD36 expression in the peritoneal macrophages of ApoE-/-:PKCθ-/- mice, and it had no effect on foam cell formation. In addition, the aortic root cross-sections of Western diet-fed ApoE-/- mice showed increased Pyk2, Gab1, PKCθ, and ATF2 phosphorylation and CD36 expression as compared with ApoE-/-:PKCθ-/- mice. Furthermore, although the monocytes from peripheral blood and the aorta of Western diet-fed ApoE-/- mice were found to contain more of Ly6Chi cells than Ly6Clo cells, the monocytes from Western diet-fed ApoE-/-:PKCθ-/- mice were found to contain more Ly6Clo cells than Ly6Chi cells. It is interesting to note that the Ly6Chi cells showed higher CD36 expression with enhanced capacity to form foam cells as compared with Ly6Clo cells. CONCLUSIONS: These findings reveal for the first time that thrombin-mediated Par1-Gα12 signaling via targeting Pyk2-Gab1-PKCθ-ATF2-dependent CD36 expression might be playing a crucial role in diet-induced atherogenesis.


Asunto(s)
Factor de Transcripción Activador 2/metabolismo , Aterosclerosis/patología , Antígenos CD36/metabolismo , Proteína Quinasa C-theta/metabolismo , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/metabolismo , Aterosclerosis/veterinaria , Antígenos CD36/antagonistas & inhibidores , Antígenos CD36/genética , Diferenciación Celular/efectos de los fármacos , Células Espumosas/citología , Células Espumosas/metabolismo , Quinasa 2 de Adhesión Focal/antagonistas & inhibidores , Quinasa 2 de Adhesión Focal/genética , Quinasa 2 de Adhesión Focal/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Expresión Génica/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Proteína Quinasa C-theta/deficiencia , Proteína Quinasa C-theta/genética , Células RAW 264.7 , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Trombina/farmacología , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
3.
J Hepatol ; 68(3): 493-504, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29080810

RESUMEN

BACKGROUND & AIMS: Hepatic stellate cells (HSCs) have a role in liver fibrosis. Guanine nucleotide-binding α-subunit 12 (Gα12) converges signals from G-protein-coupled receptors whose ligand levels are elevated in the environment during liver fibrosis; however, information is lacking on the effect of Gα12 on HSC trans-differentiation. This study investigated the expression of Gα12 in HSCs and the molecular basis of the effects of its expression on liver fibrosis. METHODS: Gα12 expression was assessed by immunostaining, and immunoblot analyses of mouse fibrotic liver tissues and primary HSCs. The role of Gα12 in liver fibrosis was estimated using a toxicant injury mouse model with Gα12 gene knockout and/or HSC-specific Gα12 delivery using lentiviral vectors, in addition to primary HSCs and LX-2 cells using microRNA (miR) inhibitors, overexpression vectors, or adenoviruses. miR-16, Gα12, and LC3 were also examined in samples from patients with fibrosis. RESULTS: Gα12 was overexpressed in activated HSCs and fibrotic liver, and was colocalised with desmin. In a carbon tetrachloride-induced fibrosis mouse model, Gα12 ablation prevented increases in fibrosis and liver injury. This effect was attenuated by HSC-specific lentiviral delivery of Gα12. Moreover, Gα12 activation promoted autophagy accompanying c-Jun N-terminal kinase-dependent ATG12-5 conjugation. In addition, miR-16 was found to be a direct inhibitor of the de novo synthesis of Gα12. Modulations of miR-16 altered autophagy in HSCs. In a fibrosis animal model or patients with severe fibrosis, miR-16 levels were lower than in their corresponding controls. Consistently, cirrhotic patient liver tissues showed Gα12 and LC3 upregulation in desmin-positive areas. CONCLUSIONS: miR-16 dysregulation in HSCs results in Gα12 overexpression, which activates HSCs by facilitating autophagy through ATG12-5 formation. This suggests that Gα12 and its regulatory molecules could serve as targets for the amelioration of liver fibrosis. LAY SUMMARY: Guanine nucleotide-binding α-subunit 12 (Gα12) is upregulated in activated hepatic stellate cells (HSCs) as a consequence of the dysregulation of a specific microRNA that is abundant in HSCs, facilitating the progression of liver fibrosis. This event is mediated by c-Jun N-terminal kinase-dependent ATG12-5 formation and the promotion of autophagy. We suggest that Gα12 and its associated regulators could serve as new targets in HSCs for the treatment of liver fibrosis.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Células Estrelladas Hepáticas/metabolismo , Cirrosis Hepática , MicroARNs/metabolismo , Animales , Autofagia/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Subunidades alfa de la Proteína de Unión al GTP G12-G13/antagonistas & inhibidores , Regulación de la Expresión Génica , Humanos , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Ratones , Inhibidor 1 de Activador Plasminogénico/metabolismo , Inhibidor 1 de Activador Plasminogénico/farmacología , Inhibidores de Serina Proteinasa/farmacología , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba
4.
Oncotarget ; 5(20): 9626-40, 2014 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-25275299

RESUMEN

Oral squamous cell carcinoma (OSCC) has a propensity to spread to the cervical lymph nodes (LN). The presence of cervical LN metastases severely impacts patient survival, whereby the two-year survival for oral cancer patients with involved LN is ~30% compared to over 80% in patients with non-involved LN. Elucidation of key molecular mechanisms underlying OSCC metastasis may afford an opportunity to target specific genes, to prevent the spread of OSCC and to improve patient survival. In this study, we demonstrated that expression of the heterotrimeric G-protein alpha-12 (Gα12) is highly up-regulated in primary tumors and LN of OSCC patients, as assessed by quantitative polymerase chain reaction (qPCR) and immunohistochemistry (IHC). We also found that exogenous expression of the constitutively activated-form of Gα12 promoted cell migration and invasion in OSCC cell lines. Correspondingly, inhibition of Gα12 expression by shRNA consistently inhibited OSCC cell migration and invasion in vitro. Further, the inhibition of G12 signaling by regulator of G-protein signaling (RGS) inhibited Gα12-mediated RhoA activation, which in turn resulted in reduced LN metastases in a tongue-orthotopic xenograft mouse model of oral cancer. This study provides a rationale for future development and evaluation of drug candidates targeting Gα12-related pathways for metastasis prevention.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Animales , Carcinoma de Células Escamosas/genética , Movimiento Celular/fisiología , Femenino , Subunidades alfa de la Proteína de Unión al GTP G12-G13/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/genética , Humanos , Metástasis Linfática , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Neoplasias de la Boca/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello , Activación Transcripcional , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Am J Physiol Cell Physiol ; 300(5): C1181-92, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21289285

RESUMEN

Enhanced vascular arginase activity impairs endothelium-dependent vasorelaxation by decreasing l-arginine availability to endothelial nitric oxide (NO) synthase, thereby reducing NO production. Elevated angiotensin II (ANG II) is a key component of endothelial dysfunction in many cardiovascular diseases and has been linked to elevated arginase activity. We determined signaling mechanisms by which ANG II increases endothelial arginase function. Results show that ANG II (0.1 µM, 24 h) elevates arginase activity and arginase I expression in bovine aortic endothelial cells (BAECs) and decreases NO production. These effects are prevented by the arginase inhibitor BEC (100 µM). Blockade of ANG II AT(1) receptors or transfection with small interfering RNA (siRNA) for Gα12 and Gα13 also prevents ANG II-induced elevation of arginase activity, but siRNA for Gαq does not. ANG II also elevates active RhoA levels and induces phosphorylation of p38 MAPK. Inhibitors of RhoA activation (simvastatin, 0.1 µM) or Rho kinase (ROCK) (Y-27632, 10 µM; H1152, 0.5 µM) block both ANG II-induced elevation of arginase activity and phosphorylation of p38 MAPK. Furthermore, pretreatment of BAECs with p38 inhibitor SB-202190 (2 µM) or transfection with p38 MAPK siRNA prevents ANG II-induced increased arginase activity/expression and maintains NO production. Additionally, inhibitors of p38 MAPK (SB-203580, 5 µg·kg(-1)·day(-1)) or arginase (ABH, 8 mg·kg(-1)·day(-1)) or arginase gene knockout in mice prevents ANG II-induced vascular endothelial dysfunction and associated enhancement of arginase. These results indicate that ANG II increases endothelial arginase activity/expression through Gα12/13 G proteins coupled to AT(1) receptors and subsequent activation of RhoA/ROCK/p38 MAPK pathways leading to endothelial dysfunction.


Asunto(s)
Angiotensina II/fisiología , Arginasa/fisiología , Endotelio Vascular/fisiopatología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Quinasas Asociadas a rho/fisiología , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Amidas/farmacología , Angiotensina II/farmacología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Arginasa/antagonistas & inhibidores , Ácidos Borónicos/farmacología , Bovinos , Línea Celular , Células Endoteliales , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/enzimología , Inhibidores Enzimáticos/farmacología , Subunidades alfa de la Proteína de Unión al GTP G12-G13/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/antagonistas & inhibidores , Imidazoles/farmacología , Ratones , Fosforilación , Piridinas/farmacología , ARN Interferente Pequeño/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Simvastatina/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Quinasas Asociadas a rho/antagonistas & inhibidores
6.
Carcinogenesis ; 31(7): 1230-7, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20478922

RESUMEN

Bortezomib is a proteasome inhibitor approved for anticancer therapy. However, variable sensitivity of tumor cells exists in this therapy probably due to differences in the expression of proteasome subunits. G(alpha)(12/13) serves modulators or signal transducers in diverse pathways. This study investigated whether cancer cells display differential sensitivity to bortezomib with reference to G(alpha)(12/13) expression, and if so, whether G(alpha)(12/13) affects the expression of proteasome subunits and their activities. Bortezomib treatment exhibited greater sensitivities in Huh7 and SNU886 cells (epithelial type) than SK-Hep1 and SNU449 cells (mesenchymal type) that exhibited higher levels of G(alpha)(12/13). Overexpression of an active mutant of G(alpha)(12) (Galpha(12)QL) or G(alpha)(13) (G(alpha)(13)QL) diminished the ability of bortezomib to induce cytotoxicity in Huh7 cells. Moreover, transfection with the minigene that disturbs G protein-coupled receptor-G protein coupling (CT12 or CT13) increased it in SK-Hep1 cells. Consistently, MiaPaCa2 cells transfected with CT12 or CT13 exhibited a greater sensitivity to bortezomib. Evidence of G(alpha)(12/13)'s antagonism on the anticancer effect of bortezomib was verified in the reversal by G(alpha)(12)QL or G(alpha)(13)QL of the minigenes' enhancement of cytotoxity. Real-time polymerase chain reaction assay enabled us to identify PSMB5, multicatalytic endopeptidase complex-like-1, and proteasome activator subunit-1 repression by CT12 or CT13. Furthermore, G(alpha)(12/13) inhibition enhanced the ability of bortezomib to repress PSMB5, as shown by immunoblotting and proteasome activity assay. Moreover, this inhibitory effect on PSMB5 was attenuated by G(alpha)G(alpha)(12)QL or G(alpha)(13)QL. In conclusion, the inhibition of G(alpha)(12/13) activities may enhance the anticancer effect of bortezomib through PSMB5 repression, providing insight into the G(alpha)(12/13) pathway for the regulation of proteasomal activity.


Asunto(s)
Antineoplásicos/farmacología , Ácidos Borónicos/farmacología , Subunidades alfa de la Proteína de Unión al GTP G12-G13/fisiología , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasoma , Pirazinas/farmacología , Bortezomib , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Línea Celular , Subunidades alfa de la Proteína de Unión al GTP G12-G13/antagonistas & inhibidores , Humanos , Etiquetado Corte-Fin in Situ , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Complejo de la Endopetidasa Proteasomal/genética , ARN Mensajero/análisis
7.
Am J Physiol Gastrointest Liver Physiol ; 297(4): G641-54, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19679818

RESUMEN

P2Y5 is a G protein-coupled receptor that binds and is activated by lysophosphatidic acid (LPA). We determined that P2Y5 transcript is expressed along the intestinal mucosa and investigated the intracellular pathways induced by P2Y5 activation, which could contribute to LPA effects on intestinal cell adhesion. P2Y5 heterologously expressed in CHO and small intestinal hBRIE 380i cells was activated by LPA resulting in an increase in intracellular calcium ([Ca(2+)](i)) when the cells concurrently expressed G(alpha)(Delta6qi5myr). P2Y5 activation also increased the phosphorylation of ERK1/2 that was sensitive to pertussis toxin. Together these indicate that P2Y5 activation by LPA induces an increase in [Ca(2+)](i) and ERK1/2 phosphorylation through G(alpha)(i). We discovered that P2Y5 was activated by farnesyl pyrophosphate (FPP) without a detectable change in [Ca(2+)](i). The activation of P2Y5 by LPA or FPP induced the activity of a serum response element (SRE)-linked luciferase reporter that was inhibited by the RGS domain of p115RhoGEF, C3 exotoxin, and Y-27632, suggesting the involvement of G(alpha)(12/13), Rho GTPase, and ROCK, respectively. However, only LPA-mediated induction of SRE reporter activity was sensitive to inhibitors targeting p38 MAPK, PI3K, PLC, and PKC. In addition, only LPA transactivated the epidermal growth factor receptor, leading to an induction of ERK1/2 phosphorylation. These observations correlate with our subsequent finding that P2Y5 activation by LPA, and not FPP, reduced intestinal cell adhesion. This study elucidates a mechanism whereby LPA can act as a luminal and/or serosal cue to alter mucosal integrity.


Asunto(s)
Adhesión Celular , Células Epiteliales/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Mucosa Intestinal/metabolismo , Lisofosfolípidos/metabolismo , Receptores Purinérgicos P2/metabolismo , Animales , Células CHO , Señalización del Calcio , Adhesión Celular/efectos de los fármacos , Cricetinae , Cricetulus , Regulación hacia Abajo , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Receptores ErbB/metabolismo , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células HeLa , Humanos , Mucosa Intestinal/efectos de los fármacos , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/metabolismo , Masculino , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Fosfatos de Poliisoprenilo/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor Cross-Talk , Receptores Purinérgicos P2/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho , Elemento de Respuesta al Suero , Sesquiterpenos/metabolismo , Transfección , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/metabolismo
8.
FASEB J ; 22(8): 2821-31, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18367648

RESUMEN

G alpha12 constitutes, along with G alpha13, one of the four families of alpha subunits of heterotrimeric G proteins. We found that the N terminus of G alpha12, but not those of other G alpha subunits, contains a predicted mitochondrial targeting sequence. Using confocal microscopy and cell fractionation, we demonstrated that up to 40% of endogenous G alpha12 in human umbilical vein endothelial cells colocalize with mitochondrial markers. N-terminal sequence of G alpha12 fused to GFP efficiently targeted the fusion protein to mitochondria. G alpha12 with mutated mitochondrial targeting sequence was still located in mitochondria, suggesting the existence of additional mechanisms for mitochondrial localization. Lysophosphatidic acid, one of the known stimuli transduced by G alpha12/13, inhibited mitochondrial motility, while depletion of endogenous G alpha12 increased mitochondrial motility. G alpha12Q229L variants uncoupled from RhoGEFs (but not fully functional activated G alpha12Q229L) induced transformation of the mitochondrial network into punctate mitochondria and resulted in a loss of mitochondrial membrane potential. All examined G alpha12Q229L variants reduced phosphorylation of Bcl-2 at Ser-70, while only mutants unable to bind RhoGEFs also decreased cellular levels of Bcl-2. These G alpha12 mutants were also more efficient Hsp90 interactors. These findings are the first demonstration of a heterotrimeric G protein alpha subunit specifically targeted to mitochondria and involved in the control of mitochondrial morphology and dynamics.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Mitocondrias/metabolismo , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Células Endoteliales/metabolismo , Células Endoteliales/ultraestructura , Subunidades alfa de la Proteína de Unión al GTP G12-G13/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Potencial de la Membrana Mitocondrial , Movimiento , Mutagénesis Sitio-Dirigida , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas c-bcl-2/química , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Interferente Pequeño/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho
9.
J Cell Sci ; 121(Pt 6): 814-24, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18285450

RESUMEN

The polarized functions of epithelia require an intact tight junction (TJ) to restrict paracellular movement and to separate membrane proteins into specific domains. TJs contain scaffolding, integral membrane and signaling proteins, but the mechanisms that regulate TJs and their assembly are not well defined. Galpha12 (GNA12) binds the TJ protein ZO-1 (TJP1), and Galpha12 activates Src to increase paracellular permeability via unknown mechanisms. Herein, we identify Src as a component of the TJ and find that recruitment of Hsp90 to activated Galpha12 is necessary for signaling. TJ integrity is disrupted by Galpha12-stimulated Src phosphorylation of ZO-1 and ZO-2 (TJP2); this phosphorylation leads to dissociation of occludin and claudin 1 from the ZO-1 protein complex. Inhibiting Hsp90 with geldanamycin blocks Galpha12-stimulated Src activation and phosphorylation, but does not affect protein levels or the Galpha12-ZO-1 interaction. Using the calcium-switch model of TJ assembly and GST-TPR (GST-fused TPR domain of PP5) pull-downs of activated Galpha12, we demonstrate that switching to normal calcium medium activates endogenous Galpha12 during TJ assembly. Thrombin increases permeability and delays TJ assembly by activating Galpha12, but not Galpha13, signaling pathways. These findings reveal an important role for Galpha12, Src and Hsp90 in regulating the TJ in established epithelia and during TJ assembly.


Asunto(s)
Células Epiteliales/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Uniones Estrechas/metabolismo , Animales , Línea Celular , Permeabilidad de la Membrana Celular , Claudina-1 , Perros , Células Epiteliales/enzimología , Subunidades alfa de la Proteína de Unión al GTP G12-G13/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Mutación , Ocludina , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Transducción de Señal , Uniones Estrechas/enzimología , Tirosina/metabolismo , Proteína de la Zonula Occludens-1 , Proteína de la Zonula Occludens-2
10.
J Vasc Surg ; 46(6): 1253-1259, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18155002

RESUMEN

BACKGROUND: Sphingosine-1-phosphate (S-1-P) is a bioactive sphingolipid that stimulates the migration of vascular smooth muscle cell (VSMC) through G-protein coupled receptors; it has been shown to activate reduced nicotinamide dinucleotide phosphate hydrogen (NAD[P]H) oxidase. The role of phospholipase C (PLC) in oxygen free radical generation, and the regulation of VSMC migration in response to S-1-P, are poorly understood. METHODS: Rat arterial VSMC were cultured in vitro. Oxygen free radical generation was measured by fluorescent redox indicator assays in response to S-1-P (0.1microM) in the presence and absence of the active PLC inhibitor (U73122; U7, 10nM) or its inactive analog U73343 (InactiveU7, 10nM). Activation of PLC was assessed by immunoprecipitation and Western blotting for the phosphorylated isozymes (beta and gamma). Small interfering (si) RNA to the G-proteins Galphai, Galphaq, and Galpha12/13 was used to downregulate specific proteins. Statistics were by one-way analysis of variance (n = 6). RESULTS: S-1-P induced time-dependent activation of PLC-beta and PLC-gamma; PLC-beta but not PLC-gamma activation was blocked by U7 but not by InactiveU7. PLC-beta activation was Galphai-independent (not blocked by pertussis toxin, a Galphai inhibitor, or Galphai2 and Galphai3 siRNA) and Galphaq-independent (not blocked by glycoprotein [GP] 2A, a Galphaq inhibitor, or Galphaq siRNA). PLC-beta activation and cell migration was blocked by siRNA to Galpha12/13. Oxygen free radical generation induced by S-1-P, as measured by dihydroethidium staining, was significantly inhibited by U7 but not by InactiveU7. Inhibition of oxygen free radicals with the inhibitor diphenyleneiodonium resulted in decreased cell migration to S-1-P. VSMC mitogen-activated protein kinase activation and VSMC migration in response to S-1-P was inhibited by PLC- inhibition. CONCLUSION: S-1-P induces oxygen free radical generation through a Galpha12/13, PLC-beta-mediated mechanism that facilitates VSMC migration. To our knowledge, this is the first description of PLC-mediated oxygen free radical generation as a mediator of S-1-P VSMC migration and illustrates the need for the definition of cell signaling to allow targeted strategies in molecular therapeutics for restenosis.


Asunto(s)
Movimiento Celular , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Lisofosfolípidos/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Fosfolipasa C beta/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Esfingosina/análogos & derivados , Animales , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Activación Enzimática , Estrenos/farmacología , Subunidades alfa de la Proteína de Unión al GTP G12-G13/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/enzimología , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/metabolismo , Compuestos Onio/farmacología , Toxina del Pertussis/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Fosfolipasa C beta/antagonistas & inhibidores , Fosfolipasa C gamma/metabolismo , Fosforilación , Pirrolidinonas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Esfingosina/metabolismo , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Mol Pharmacol ; 69(6): 2068-75, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16554409

RESUMEN

Because phospholipase C epsilon (PLC-epsilon) is activated by Galpha(12/13) and Rho family GTPases, we investigated whether these G proteins contribute to the increased inositol lipid hydrolysis observed in COS-7 cells after activation of certain G protein-coupled receptors. Stimulation of inositol lipid hydrolysis by endogenous lysophosphatidic acid (LPA) or thrombin receptors was markedly enhanced by the expression of PLC-epsilon. Expression of the LPA(1) or PAR1 receptor increased inositol phosphate production in response to LPA or SFLLRN, respectively, and these agonist-stimulated responses were markedly enhanced by coexpression of PLC-epsilon. Both LPA(1) and PAR1 receptor-mediated activation of PLC-epsilon was inhibited by coexpression of the regulator of G protein signaling (RGS) domain of p115RhoGEF, a GTPase-activating protein for Galpha(12/13) but not by expression of the RGS domain of GRK2, which inhibits Galpha(q) signaling. In contrast, activation of the G(q)-coupled M1 muscarinic or P2Y(2) purinergic receptor was neither enhanced by coexpression with PLC-epsilon nor inhibited by the RGS domain of p115RhoGEF but was blocked by expression of the RGS domain of GRK2. Expression of the Rho inhibitor C3 botulinum toxin did not affect LPA- or SFLLRN-stimulated inositol lipid hydrolysis in the absence of PLC-epsilon but completely prevented the PLC-epsilon-dependent increase in inositol phosphate accumulation. Likewise, C3 toxin blocked the PLC-epsilon-dependent stimulatory effects of the LPA(1), LPA(2), LPA(3), or PAR1 receptor but had no effect on the agonist-promoted inositol phosphate response of the M1 or P2Y(2) receptor. Moreover, PLC-epsilon-dependent stimulation of inositol phosphate accumulation by activation of the epidermal growth factor receptor, which involves Ras- but not Rho-mediated activation of the phospholipase, was unaffected by C3 toxin. These studies illustrate that specific LPA and thrombin receptors promote inositol lipid signaling via activation of Galpha(12/13) and Rho.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Receptor PAR-1/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Fosfolipasas de Tipo C/metabolismo , Proteínas de Unión al GTP rho/metabolismo , ADP Ribosa Transferasas/farmacología , Animales , Toxinas Botulínicas/farmacología , Células COS , Chlorocebus aethiops , Subunidades alfa de la Proteína de Unión al GTP G12-G13/antagonistas & inhibidores , Hidrólisis , Fosfatos de Inositol/metabolismo , Metabolismo de los Lípidos , Fragmentos de Péptidos/farmacología , Fosfoinositido Fosfolipasa C , Receptores de Trombina/metabolismo , Fosfolipasas de Tipo C/antagonistas & inhibidores , Proteínas de Unión al GTP rho/agonistas , Proteínas de Unión al GTP rho/antagonistas & inhibidores
12.
Mol Pharmacol ; 69(3): 975-82, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16326932

RESUMEN

Endothelial nitric-oxide synthase (eNOS) plays a crucial role in the regulation of a variety of cardiovascular and pulmonary functions in both normal and pathological conditions. Multiple signaling inputs, including calcium, caveolin-1, phosphorylation by several kinases, and binding to the 90-kDa heat shock protein (Hsp90), regulate eNOS activity. Here, we report a novel mechanism of G protein-dependent regulation of eNOS. We demonstrate that in mammalian cells, the alpha subunit of heterotrimeric G12 protein (G alpha12) can form a complex with eNOS in an activation- and Hsp90-independent manner. Our data show that G alpha12 does not affect eNOS-specific activity, but it strongly enhances total eNOS activity by increasing cellular levels of eNOS. Experiments using inhibition of protein or mRNA synthesis show that G alpha12 increases the expression of eNOS by increasing half-life of both eNOS protein and eNOS mRNA. Small interfering RNA-mediated depletion of endogenous G alpha12 decreases eNOS levels. A quantitative correlation can be detected between the extent of down-regulation of G alpha12 and eNOS in endothelial cells after prolonged treatment with thrombin. G protein-dependent increase of eNOS expression represents a novel mechanism by which heterotrimeric G proteins can regulate the activity of downstream signaling molecules.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Animales , Células Cultivadas , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/enzimología , Activación Enzimática , Estabilidad de Enzimas , Subunidades alfa de la Proteína de Unión al GTP G12-G13/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Óxido Nítrico Sintasa de Tipo III/genética , Estabilidad del ARN , ARN Mensajero/análisis , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Trombina/farmacología
13.
J Biol Chem ; 280(18): 18434-41, 2005 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-15743761

RESUMEN

In the present study, we examined signal transduction mechanism of reactive oxygen species (ROS) production and the role of ROS in angiotensin II-induced activation of mitogen-activated protein kinases (MAPKs) in rat neonatal cardiomyocytes. Among three MAPKs, c-Jun NH(2)-terminal kinase (JNK) and p38 MAPK required ROS production for activation, as an NADPH oxidase inhibitor, diphenyleneiodonium, inhibited the activation. The angiotensin II-induced activation of JNK and p38 MAPK was also inhibited by the expression of the Galpha(12/13)-specific regulator of G protein signaling (RGS) domain, a specific inhibitor of Galpha(12/13), but not by an RGS domain specific for Galpha(q). Constitutively active Galpha(12)- or Galpha(13)-induced activation of JNK and p38 MAPK, but not extracellular signal-regulated kinase (ERK), was inhibited by diphenyleneiodonium. Angiotensin II receptor stimulation rapidly activated Galpha(13), which was completely inhibited by the Galpha(12/13)-specific RGS domain. Furthermore, the Galpha(12/13)-specific but not the Galpha(q)-specific RGS domain inhibited angiotensin II-induced ROS production. Dominant negative Rac inhibited angiotensin II-stimulated ROS production, JNK activation, and p38 MAPK activation but did not affect ERK activation. Rac activation was mediated by Rho and Rho kinase, because Rac activation was inhibited by C3 toxin and a Rho kinase inhibitor, Y27632. Furthermore, angiotensin II-induced Rho activation was inhibited by Galpha(12/13)-specific RGS domain but not dominant negative Rac. An inhibitor of epidermal growth factor receptor kinase AG1478 did not affect angiotensin II-induced JNK activation cascade. These results suggest that Galpha(12/13)-mediated ROS production through Rho and Rac is essential for JNK and p38 MAPK activation.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Miocitos Cardíacos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Angiotensina/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Antagonistas de Receptores de Angiotensina , Animales , Animales Recién Nacidos , Bencimidazoles/farmacología , Compuestos de Bifenilo , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Subunidades alfa de la Proteína de Unión al GTP G12-G13/antagonistas & inhibidores , Ratones , Miocitos Cardíacos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Tetrazoles/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...